Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Mol Ther ; 25(3): 593-605, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28190779

ABSTRACT

Recently, an engineered Homeobox-nucleoporin fusion gene, NUP98-HOXA10HD or NA10HD, was reported to expand and maintain murine hematopoietic stem cells (HSCs). We postulated that NA10HD would increase the number of human γ-globin-expressing cells to therapeutic levels. We developed a double gene lentiviral vector encoding both human γ-globin and NA10HD, which was used to transduce human peripheral blood CD34+ cells and increased engraftment 2- to 2.5-fold at 15 weeks post-transplantation in immunodeficient mice. In ß-thalassemic mice transplanted with ß-thalassemic HSCs transduced with the γ-globin/NA10HD vector, the number of fetal hemoglobin (HbF)-expressing cells was significantly increased after 3 months, leading to resolution of the anemia. Furthermore, the increases in HbF were maintained at 6 months and persisted after secondary transplantation. In addition, NA10HD enrichment of transduced HSCs led to HbF increases without affecting homeostasis of the white blood cell lineages. Our results suggest that NA10HD increases the number of γ-globin-transduced HSCs that engraft, leading to an elevated number of fetal hemoglobin-containing red cells. These effects of NA10HD provide an improved platform for testing of the therapeutic efficacy of novel globin vectors and provide further impetus to develop safe and effective methods for selective expansion of genetically modified cells.


Subject(s)
Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Lentivirus/genetics , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , beta-Thalassemia/genetics , gamma-Globins/genetics , Animals , Disease Models, Animal , Erythrocytes/cytology , Erythrocytes/metabolism , Fetal Hemoglobin/metabolism , Gene Order , Gene Transfer Techniques , Genetic Loci , Graft Survival , Hematopoietic Stem Cell Transplantation , Homeobox A10 Proteins , Humans , Mice , Transduction, Genetic , Transplantation, Heterologous , beta-Thalassemia/metabolism , beta-Thalassemia/therapy
2.
Data Brief ; 7: 1288-95, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27141526

ABSTRACT

This paper describes data related to a research article titled, "Fas-antisense long noncoding RNA is differentially expressed during maturation of human erythrocytes and confers resistance to Fas-mediated cell death" [1]. Long noncoding RNAs (lncRNAs) are increasingly appreciated for their capacity to regulate many steps of gene expression. While recent studies suggest that many lncRNAs are functional, the scope of their actions throughout human biology is largely undefined including human red blood cell development (erythropoiesis). Here we include expression data for 82 lncRNAs during early, intermediate and late stages of human erythropoiesis using a commercial qPCR Array. From these data, we identified lncRNA Fas-antisense 1 (Fas-AS1 or Saf) described in the research article. Also included are 5' untranslated sequences (UTR) for lncRNA Saf with transcription factor target sequences identified. Quantitative RT-PCR data demonstrate relative levels of critical erythroid transcription factors, GATA-1 and KLF1, in K562 human erythroleukemia cells and maturing erythroblasts derived from human CD34(+) cells. End point and quantitative RT-PCR data for cDNA prepared using random hexamers versus oligo(dT)18 revealed that lncRNA Saf is not effectively polyadenylated. Finally, we include flow cytometry histograms demonstrating Fas levels on maturing erythroblasts derived from human CD34(+) cells transduced using mock conditions or with lentivirus particles encoding for Saf.

3.
Blood Cells Mol Dis ; 58: 57-66, 2016 May.
Article in English | MEDLINE | ID: mdl-27067490

ABSTRACT

Long noncoding RNAs (lncRNAs) interact with other RNAs, DNA and/or proteins to regulate gene expression during development. Erythropoiesis is one developmental process that is tightly controlled throughout life to ensure accurate red blood cell production and oxygen transport to tissues. Thus, homeostasis is critical and maintained by competitive outcomes of pro- and anti-apoptotic pathways. LncRNAs are expressed during blood development; however, specific functions are largely undefined. Here, a culture model of human erythropoiesis revealed that lncRNA Fas-antisense 1 (Fas-AS1 or Saf) was induced during differentiation through the activity of essential erythroid transcription factors GATA-1 and KLF1. Saf was also negatively regulated by NF-κB, where decreasing NF-κB activity levels tracked with increasing transcription of Saf. Furthermore, Saf over-expression in erythroblasts derived from CD34(+) hematopoietic stem/progenitor cells of healthy donors reduced surface levels of Fas and conferred protection against Fas-mediated cell death signals. These studies reveal a novel lncRNA-regulated mechanism that modulates a critical cell death program during human erythropoiesis.


Subject(s)
Apoptosis , Erythroblasts/cytology , Erythrocytes/cytology , Erythropoiesis , RNA, Long Noncoding/genetics , fas Receptor/genetics , Cell Line, Tumor , Erythroblasts/metabolism , Erythrocytes/metabolism , GATA1 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/metabolism , fas Receptor/metabolism
4.
Oncotarget ; 7(12): 13810-26, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26885613

ABSTRACT

In multicellular organisms, cell growth and differentiation is controlled in part by programmed cell death or apoptosis. One major apoptotic pathway is triggered by Fas receptor (Fas)-Fas ligand (FasL) interaction. Neoplastic cells are frequently resistant to Fas-mediated apoptosis, evade Fas signals through down regulation of Fas and produce soluble Fas proteins that bind FasL thereby blocking apoptosis. Soluble Fas (sFas) is an alternative splice product of Fas pre-mRNA, commonly created by exclusion of transmembrane spanning sequences encoded within exon 6 (FasΔEx6). Long non-coding RNAs (lncRNAs) interact with other RNAs, DNA, and proteins to regulate gene expression. One lncRNA, Fas-antisense or Saf, was shown to participate in alternative splicing of Fas pre-mRNA through unknown mechanisms. We show that Saf is localized in the nucleus where it interacts with Fas receptor pre-mRNA and human splicing factor 45 (SPF45) to facilitate alternative splicing and exclusion of exon 6. The product is a soluble Fas protein that protects cells against FasL-induced apoptosis. Collectively, these studies reveal a novel mechanism to modulate this critical cell death program by an lncRNA and its protein partner.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation , RNA Splicing Factors/metabolism , RNA Splicing , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , fas Receptor/metabolism , HEK293 Cells , HeLa Cells , Humans , RNA Splicing Factors/genetics , fas Receptor/genetics
5.
PLoS One ; 11(1): e0147059, 2016.
Article in English | MEDLINE | ID: mdl-26761813

ABSTRACT

Techniques to expand human hematopoietic stem cells ex-vivo could be beneficial to the fields of clinical hematopoietic stem cell transplantation and gene therapy targeted at hematopoietic stem cells. NUP98-HOXA10HD is a relatively newly discovered fusion gene that in mouse transplant experiments has been shown to increase numbers of hematopoietic stem cells. We evaluated whether this fusion gene could be used to expand engrafting human primitive CD34+ cells in an immunodeficient mouse model. Gene transfer was achieved using a lentiviral based vector. The engraftment of mobilized peripheral blood human CD34+ cells grown in culture for one week after gene transfer was evaluated 3-4 months after transplant and found to be 2-3 fold higher in the NUP98-HOXA10HD groups as compared to controls. These data suggest an expansive effect at least at the short term human repopulating cell level. Further evaluation in long term repopulating models and investment in a NUP98-HOXA10HD protein seems worthy of consideration. Additionally, the results here provide strong impetus to utilize NUP98-HOXA10HD as a tool to search for underlying genes and pathways involved in hematopoietic stem cell expansion that can be enhanced and have an even more potent expansive effect.


Subject(s)
Gene Expression , Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Nuclear Pore Complex Proteins/genetics , Recombinant Fusion Proteins/genetics , Animals , Antigens, Surface/metabolism , Gene Order , Genetic Vectors/genetics , Homeobox A10 Proteins , Homeodomain Proteins/chemistry , Humans , Immunophenotyping , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Lentivirus/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Models, Animal , Transduction, Genetic
6.
Mol Ther Methods Clin Dev ; 2: 15045, 2015.
Article in English | MEDLINE | ID: mdl-26665131

ABSTRACT

Patients with severe sickle cell disease (SCD) are candidates for gene therapy using autologous hematopoietic stem cells (HSCs), but concomitant multi-organ disease may contraindicate pretransplant conditioning with full myeloablation. We tested whether nonmyeloablative conditioning, a regimen used successfully for allogeneic bone marrow transplantation of adult SCD patients, allows engraftment of γ-globin gene-corrected cells to a therapeutic level in the Berkeley mouse model of SCD. Animals transplanted according to this regimen averaged 35% engraftment of transduced hematopoietic stem cells with an average vector copy < 2.0. Fetal hemoglobin (HbF) levels ranged from 20 to 44% of total hemoglobin and approximately two-thirds of circulating red blood cells expressed HbF detected by immunofluorescence (F-cells). Gene therapy treatment of SCD mice ameliorated anemia, reduced hyperleukocytosis, improved renal function, and reduced iron accumulation in liver, spleen, and kidneys. Thus, modest levels of chimerism with donor cells expressing high levels of HbF from an insulated γ-globin lentiviral vector can improve the pathology of SCD in mice, thereby illustrating a potentially safe and effective strategy for gene therapy in humans.

7.
Exp Hematol ; 43(7): 565-77.e1-10, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25907033

ABSTRACT

The transcription factor GATA2 is highly expressed in hematopoietic stem cells and is downregulated during lineage maturation. Gain of function mutations, loss of function mutations, and overexpression of GATA2 have been reported in acute myeloid leukemia. In previous studies, we and others showed that GATA2 overexpression at high levels, similar to that seen in hematopoietic stem cells, blocked differentiation of hematopoietic stem cells and progenitors. To better understand the effects of GATA2, we designed a Tamoxifen-inducible GATA2-estrogen receptor (ERT) vector. In the absence of Tamoxifen, small amounts of GATA2-ERT were still able to enter the nucleus in mouse bone marrow (BM) cells, providing us with a tool to test the effects of low-level GATA2 overexpression. We observed that this low-level GATA2 overexpression enhanced self-renewal of myeloid progenitors in vitro and resulted in immortalization of BM cells to myeloid cell lines. Continuous GATA2-ERT expression was required for the proliferation of these immortalized lines. Myeloid expansion and a block in T and B lineage differentiation were observed in mice transplanted with GATA2-ERT-expressing BM cells. Myeloid expansion occurred after the granulocyte monocyte progenitor stage, and lymphoid block was distal to the common lymphoid progenitor in transgenic mice. GATA2 appeared to induce growth via downstream activation of Nmyc and Hoxa9. Our results demonstrate that GATA2 overexpression at low level confers self-renewal capacity to myeloid progenitors and is relevant to myeloid leukemia development.


Subject(s)
Bone Marrow Cells/pathology , Cell Transformation, Neoplastic/genetics , GATA2 Transcription Factor/physiology , Gene Expression Regulation, Leukemic , Lymphopoiesis/genetics , Myeloid Cells/pathology , Myelopoiesis/genetics , Animals , B-Lymphocytes/pathology , Bone Marrow Cells/metabolism , Cell Division , Cell Nucleus/metabolism , Cells, Cultured , Colony-Forming Units Assay , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Leukemic/drug effects , Genes, Synthetic , Genes, myc , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Myeloid Cells/metabolism , Receptors, Estrogen/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/pathology , Tamoxifen/pharmacology
8.
Exp Hematol ; 43(5): 346-351, 2015 May.
Article in English | MEDLINE | ID: mdl-25681747

ABSTRACT

Sickle cell disease (SCD) can be cured by allogeneic hematopoietic stem cell transplant. However, this is only possible when a matched donor is available, making the development of gene therapy using autologous hematopoietic stem cells a highly desirable alternative. We used a culture model of human erythropoiesis to directly compare two insulated, self-inactivating, and erythroid-specific lentiviral vectors, encoding for γ-globin (V5m3-400) or a modified ß-globin (ßAS3-FB) for production of antisickling hemoglobin (Hb) and correction of red cell deformability after deoxygenation. Bone marrow CD34+ cells from three SCD patients were transduced using V5m3-400 or ßAS3-FB and compared with mock-transduced SCD or healthy donor CD34+ cells. Lentiviral transduction did not impair cell growth or differentiation, as gauged by proliferation and acquisition of erythroid markers. Vector copy number averaged approximately one copy per cell, and corrective globin mRNA levels were increased more than sevenfold over mock-transduced controls. Erythroblasts derived from healthy donor and mock-transduced SCD cells produced a low level of fetal Hb that was increased to 23.6 ± 4.1% per vector copy for cells transduced with V5m3-400. Equivalent levels of modified normal adult Hb of 17.6 ± 3.8% per vector copy were detected for SCD cells transduced with ßAS3-FB. These levels of antisickling Hb production were sufficient to reduce sickling of terminal-stage red blood cells upon deoxygenation. We concluded that the achieved levels of fetal Hb and modified normal adult Hb would likely prove therapeutic to SCD patients who lack matched donors.


Subject(s)
Bone Marrow Cells/metabolism , Lentivirus/genetics , beta-Globins/genetics , gamma-Globins/genetics , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/therapy , Antigens, CD34/metabolism , Fetal Hemoglobin/genetics , Flow Cytometry , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Hemoglobins/genetics , Humans , Reverse Transcriptase Polymerase Chain Reaction , beta-Globins/metabolism , gamma-Globins/metabolism
9.
Mol Ther Methods Clin Dev ; 1: 14020, 2014.
Article in English | MEDLINE | ID: mdl-26015964

ABSTRACT

Foamy virus (FV) vectors are promising tools for gene therapy, but low titer is a major challenge for large-scale clinical trials. Here, we increased FV vector titer 50-fold by constructing novel vector plasmids and using polyethylenimine-mediated transfection. FV and lentiviral (LV) vectors were used separately to transduce human CD34(+) cells at multiplicities of infection of 25, and those cells were transplanted into immunodeficient mice. FV vector transduction frequencies of repopulating human cells were 37.1 ± 1.9% in unstimulated cells and 36.9 ± 2.2% in prestimulated cells, and engraftment frequencies were 40.9 ± 4.9% in unstimulated cells and 47.1 ± 3.3% in prestimulated cells. Engraftment frequencies of FV vector-transduced cells were significantly higher than those of LV vector-transduced cells. Linear amplification-mediated PCR with Illumina paired-end runs showed that all human chromosomes contained FV provirus. FV had an integration preference near transcriptional start sites and CpG islands of RefSeq genes but not within genes. Repopulating lymphoid and myeloid cells contained common integration sites, suggesting that FV vector could transduce multilineage hematopoietic stem/progenitor populations. Our new FV vector backbone may be a suitable candidate for developing therapeutic FV vectors for use in clinical trials.

10.
Blood ; 122(17): 2987-96, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24041575

ABSTRACT

Hematopoietic stem cells are both necessary and sufficient to sustain the complete blood system of vertebrates. Here we show that Nfix, a member of the nuclear factor I (Nfi) family of transcription factors, is highly expressed by hematopoietic stem and progenitor cells (HSPCs) of murine adult bone marrow. Although short hairpin RNA-mediated knockdown of Nfix expression in Lineage(-)Sca-1(+)c-Kit(+) HSPCs had no effect on in vitro cell growth or viability, Nfix-depleted HSPCs displayed a significant loss of colony-forming potential, as well as short- and long-term in vivo hematopoietic repopulating activity. Analysis of recipient mice at 4 to 20 days posttransplant revealed that Nfix-depleted HSPCs are established in the bone marrow, but fail to persist due to increased apoptotic cell death. Gene expression profiling of Nfix-depleted HSPCs reveals that loss of Nfix expression in HSPCs is concomitant with a decrease in the expression of multiple genes known to be important for HSPCs survival, such as Erg, Mecom, and Mpl. These data reveal that Nfix is a novel regulator of HSPCs survival posttransplantation and establish a role for Nfi genes in the regulation of this cellular compartment.


Subject(s)
Adult Stem Cells/metabolism , Bone Marrow Cells/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , NFI Transcription Factors/genetics , Adult Stem Cells/cytology , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Apoptosis , Bone Marrow Cells/cytology , Cell Survival , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NFI Transcription Factors/deficiency , NFI Transcription Factors/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Signal Transduction , Transcription Factors , Transcriptional Regulator ERG
11.
Article in English | MEDLINE | ID: mdl-23125203

ABSTRACT

Retroviral vector-mediated gene transfer into hematopoietic stem cells provides a potentially curative therapy for severe ß-thalassemia. Lentiviral vectors based on human immunodeficiency virus have been developed for this purpose and have been shown to be effective in curing thalassemia in mouse models. One participant in an ongoing clinical trial has achieved transfusion independence after gene transfer into bone marrow stem cells owing, in part, to a genetically modified, dominant clone. Ongoing efforts are focused on improving the efficiency of lentiviral vector-mediated gene transfer into stem cells so that the curative potential of gene transfer can be consistently achieved.


Subject(s)
Genetic Therapy , Genetic Vectors , Globins/genetics , Thalassemia/therapy , Animals , Clinical Trials as Topic , Disease Models, Animal , Gammaretrovirus , Genetic Vectors/adverse effects , Humans , Lentivirus , Locus Control Region , Stem Cells
12.
Mol Ther ; 20(10): 1882-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22871664

ABSTRACT

Human immunodeficiency virus type 1 (HIV1) vectors poorly transduce rhesus hematopoietic cells due to species-specific restriction factors, including the tripartite motif-containing 5 isoformα (TRIM5α) which targets the HIV1 capsid. We previously developed a chimeric HIV1 (χHIV) vector system wherein the vector genome is packaged with the simian immunodeficiency virus (SIV) capsid for efficient transduction of both rhesus and human CD34(+) cells. To evaluate whether χHIV vectors could efficiently transduce rhesus hematopoietic repopulating cells, we performed a competitive repopulation assay in rhesus macaques, in which half of the CD34(+) cells were transduced with standard SIV vectors and the other half with χHIV vectors. As compared with SIV vectors, χHIV vectors achieved higher vector integration, and the transgene expression rates were two- to threefold higher in granulocytes and red blood cells and equivalent in lymphocytes and platelets for 2 years. A recipient of χHIV vector-only transduced cells reached up to 40% of transgene expression rates in granulocytes and lymphocytes and 20% in red blood cells. Similar to HIV1 and SIV vectors, χHIV vector frequently integrated into gene regions, especially into introns. In summary, our χHIV vector demonstrated efficient transduction for rhesus long-term repopulating cells, comparable with SIV vectors. This χHIV vector should allow preclinical testing of HIV1-based therapeutic vectors in large animal models.


Subject(s)
Genetic Vectors/genetics , HIV-1/genetics , Hematopoietic Stem Cells , Transduction, Genetic , Animals , Antigens, CD34/metabolism , Blotting, Southern , Capsid/metabolism , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Line , Hematopoietic Stem Cell Transplantation , Humans , Macaca mulatta , Real-Time Polymerase Chain Reaction , Simian Immunodeficiency Virus/genetics , Transgenes
13.
Mol Ther ; 20(6): 1158-66, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22434139

ABSTRACT

Foamy viruses (FVs) (spumaretroviruses) are good alternative to retroviruses as gene therapy vector. Despite four decades since the discovery of FV, its receptor molecule is still unknown. FV vector transduction of human CD34(+) cells was inhibited by culture with fibronectin. Because fibronectin contains heparin-binding domain, the interactions of fibronectin with heparan sulfate (HS) on cells might be inhibitory to FV transduction. These observations led us to investigate whether HS is a receptor for FV. Two mutant CHO cell lines (but not parental wild type) lacking cell surface HS but not chondroitin sulfate (CS) were largely resistant to FV attachment and transduction. Inhibition of HS expression using enzymes or chemicals greatly reduced FV transduction in human, monkey, and rodent cells. Raji cells, which lack HS and were largely resistant to FV, were rendered more permissive through ectopic expression of syndecan-1, which contains HS. In contrast, mutant syndecan-1-expressing cells were largely resistant to FV. Our findings indicate that cellular HS is a receptor for FV. Identifying FV receptor will enable better understanding of its entry process and optimal use as gene therapy vector to treat inherited and pathogenic diseases.


Subject(s)
Cell Membrane/chemistry , Heparitin Sulfate/metabolism , Receptors, Virus/metabolism , Spumavirus/metabolism , Animals , CHO Cells , Cell Line , Cell Membrane/metabolism , Cricetinae , Down-Regulation , Fibronectins/metabolism , Gene Expression , Haplorhini , Heparitin Sulfate/biosynthesis , Humans , Mice , Polysaccharide-Lyases/metabolism , Rodentia , Syndecan-1/genetics , Syndecan-1/metabolism , Transduction, Genetic
14.
Blood ; 119(8): 1915-21, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22130804

ABSTRACT

Sickle cell anemia is characterized by chronic hemolysis coupled with extensive vascular inflammation. This inflammatory state also mechanistically promotes a high risk of lethal, invasive pneumococcal infection. Current treatments to reduce vaso-occlusive complications include chronic hydroxyurea therapy to induce fetal hemoglobin. Because hydroxyurea also reduces leukocytosis, an understanding of the impact of this treatment on pneumococcal pathogenesis is needed. Using a sickle cell mouse model of pneumococcal pneumonia and sepsis, administration of hydroxyurea was found to significantly improve survival. Hydroxyurea treatment decreased neutrophil extravasation into the infected lung coincident with significantly reduced levels of E-selectin in serum and on pulmonary epithelia. The protective effect of hydroxyurea was abrogated in mice deficient in E-selectin. The decrease in E-selectin levels was also evident in human sickle cell patients receiving hydroxyurea therapy. These data indicate that in addition to induction of fetal hemoglobin, hydroxyurea attenuates leukocyte-endothelial interactions in sickle cell anemia, resulting in protection against lethal pneumococcal sepsis.


Subject(s)
Anemia, Sickle Cell/drug therapy , E-Selectin/metabolism , Hydroxyurea/therapeutic use , Pneumonia, Pneumococcal/prevention & control , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/metabolism , Animals , Antisickling Agents/therapeutic use , Child , Disease Models, Animal , E-Selectin/blood , E-Selectin/genetics , Female , Humans , Immunohistochemistry , Lung/drug effects , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Neutrophils/drug effects , Neutrophils/pathology , Pneumonia, Pneumococcal/complications , Survival Analysis
15.
Clin Immunol ; 141(2): 169-76, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21865090

ABSTRACT

FOXP3 is critical for the development and function of CD4(+)CD25(bright) natural regulatory T cells (nTreg). Individuals harboring mutations in FOXP3 develop immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX). We describe a child diagnosed with IPEX who underwent a reduced intensity, T and B cell depleted, matched unrelated donor bone marrow transplant followed by clinical resolution. Using lineage-specific donor chimerism studies, we demonstrate that non-myeloablative HSCT resolves disease in the context of low level donor hematopoietic stem cell (HSC) engraftment. Despite low-levels of donor HSC, thymically-derived nTreg and to a lesser extent CD4(+) and CD8(+) T cells, exhibit a selective in vivo growth advantage for populations containing a functional FOXP3 gene. Moreover, nTreg from this patient show regulatory function directly ex vivo. These results have implications for improving clinical therapy for patients with IPEX and provide mechanistic insight into the in vivo development of human nTreg and unexpectedly, non-regulatory T cells.


Subject(s)
Bone Marrow Transplantation , Forkhead Transcription Factors/deficiency , Genetic Diseases, X-Linked/surgery , Immunologic Deficiency Syndromes/surgery , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Transplantation Conditioning/methods , Alemtuzumab , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm , Cell Survival , Forkhead Transcription Factors/genetics , Genetic Diseases, X-Linked/genetics , Graft Enhancement, Immunologic , Graft Survival , Humans , Immunologic Deficiency Syndromes/genetics , Infant , Interleukin-2 Receptor alpha Subunit/analysis , Lymphocyte Depletion , Male , Melphalan , Point Mutation , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Regulatory/cytology , Thiotepa , Transplantation, Homologous , Vidarabine/analogs & derivatives
16.
Hum Gene Ther ; 22(6): 689-96, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21275758

ABSTRACT

Children with leukocyte adhesion deficiency type 1 (LAD-1) and dogs with canine LAD (CLAD) develop life-threatening bacterial infections due to mutations in the leukocyte integrin CD18. Here, we compared the human phosphoglycerate kinase (hPGK) promoter to the murine stem cell virus (MSCV) promoter/enhancer in a self-inactivating HIV-1-derived lentiviral vector to treat animals with CLAD. Four CLAD dogs were infused with CD34(+) cells transduced with the hPGK vector, and two CLAD dogs received MSCV vector-transduced CD34(+) cells. Infusions were preceded by a nonmyeloablative dose of 200 cGy total body irradiation. Comparable numbers of transduced cells were infused in each group of animals. Only one of four CLAD animals treated with the hPGK-cCD18 vector had reversal of CLAD, whereas both MSCV-cCD18 vector-treated dogs had reversal of the phenotype. Correction of CLAD depends both upon the percentage of CD18(+) myeloid cells and the level of expression of CD18 on individual myeloid cells. In this regard, the hPGK promoter directed low levels of expression of CD18 on neutrophils compared to the MSCV promoter, likely contributing to the suboptimal clinical outcome with the hPGK vector.


Subject(s)
CD18 Antigens/genetics , Dog Diseases/therapy , Genetic Therapy/methods , Leukocyte-Adhesion Deficiency Syndrome/therapy , Leukocyte-Adhesion Deficiency Syndrome/veterinary , Animals , Dogs , Genetic Vectors , HIV-1/genetics , Humans , Lentivirus/genetics , Mice , Phosphoglycerate Kinase/genetics , Promoter Regions, Genetic , Stem Cells , Transduction, Genetic/methods , Whole-Body Irradiation
17.
Blood ; 117(15): 3945-53, 2011 Apr 14.
Article in English | MEDLINE | ID: mdl-21321359

ABSTRACT

In humans, embryonic, fetal, and adult hemoglobins are sequentially expressed in developing erythroblasts during ontogeny. For the past 40 years, this process has been the subject of intensive study because of its value to enlighten the biology of developmental gene regulation and because fetal hemoglobin can significantly ameliorate the clinical manifestations of both sickle cell disease and ß-thalassemia. Understanding the normal process of loss of fetal globin expression and activation of adult globin expression could potentially lead to new therapeutic approaches for these hemoglobin disorders. Herein, we briefly review the history of the study of hemoglobin switching and then focus on recent discoveries in the field that now make new therapeutic approaches seem feasible in the future. Erythroid-specific knockdown of fetal gene repressors or enforced expression of fetal gene activators may provide clinically applicable approaches for genetic treatment of hemoglobin disorders that would benefit from increased fetal hemoglobin levels.


Subject(s)
Fetal Hemoglobin/genetics , Gene Expression Regulation, Developmental/physiology , Transcription, Genetic/physiology , Humans
19.
Blood ; 117(10): 2817-26, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21156846

ABSTRACT

ß-Thalassemia major results from severely reduced or absent expression of the ß-chain of adult hemoglobin (α2ß2;HbA). Increased levels of fetal hemoglobin (α2γ2;HbF), such as occurs with hereditary persistence of HbF, ameliorate the severity of ß-thalassemia, raising the potential for genetic therapy directed at enhancing HbF. We used an in vitro model of human erythropoiesis to assay for enhanced production of HbF after gene delivery into CD34(+) cells obtained from mobilized peripheral blood of normal adults or steady-state bone marrow from patients with ß-thalassemia major. Lentiviral vectors encoding (1) a human γ-globin gene with or without an insulator, (2) a synthetic zinc-finger transcription factor designed to interact with the γ-globin gene promoters, or (3) a short-hairpin RNA targeting the γ-globin gene repressor, BCL11A, were tested. Erythroid progeny of normal CD34(+) cells demonstrated levels of HbF up to 21% per vector copy. For ß-thalassemic CD34(+) cells, similar gene transfer efficiencies achieved HbF production ranging from 45% to 60%, resulting in up to a 3-fold increase in the total cellular Hb content. These observations suggest that both lentiviral-mediated γ-globin gene addition and genetic reactivation of endogenous γ-globin genes have potential to provide therapeutic HbF levels to patients with ß-globin deficiency.


Subject(s)
Erythroid Precursor Cells/metabolism , Fetal Hemoglobin/biosynthesis , Gene Transfer Techniques , Genetic Therapy , beta-Thalassemia/therapy , gamma-Globins/genetics , Antigens, CD34/metabolism , Blotting, Southern , Blotting, Western , Cell Separation , Erythropoiesis/physiology , Fetal Hemoglobin/genetics , Flow Cytometry , Genetic Vectors , Humans , Lentivirus/genetics , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL